#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Reengineering Redox Sensitive GFP to Measure Mycothiol Redox Potential of during Infection


Mycobacterium tuberculosis (Mtb) survives under oxidatively hostile environments encountered inside host phagocytes. To protect itself from oxidative stress, Mtb produces millimolar concentrations of mycothiol (MSH), which functions as a major cytoplasmic redox buffer. Here, we introduce a novel system for real-time imaging of mycothiol redox potential (EMSH) within Mtb cells during infection. We demonstrate that coupling of Mtb MSH-dependent oxidoreductase (mycoredoxin-1; Mrx1) to redox-sensitive GFP (roGFP2; Mrx1-roGFP2) allowed measurement of dynamic changes in intramycobacterial EMSH with unprecedented sensitivity and specificity. Using Mrx1-roGFP2, we report the first quantitative measurements of EMSH in diverse mycobacterial species, genetic mutants, and drug-resistant patient isolates. These cellular studies reveal, for the first time, that the environment inside macrophages and sub-vacuolar compartments induces heterogeneity in EMSH of the Mtb population. Further application of this new biosensor demonstrates that treatment of Mtb infected macrophage with anti-tuberculosis (TB) drugs induces oxidative shift in EMSH, suggesting that the intramacrophage milieu and antibiotics cooperatively disrupt the MSH homeostasis to exert efficient Mtb killing. Lastly, we analyze the membrane integrity of Mtb cells with varied EMSH during infection and show that subpopulation with higher EMSH are susceptible to clinically relevant antibiotics, whereas lower EMSH promotes antibiotic tolerance. Together, these data suggest the importance of MSH redox signaling in modulating mycobacterial survival following treatment with anti-TB drugs. We anticipate that Mrx1-roGFP2 will be a major contributor to our understanding of redox biology of Mtb and will lead to novel strategies to target redox metabolism for controlling Mtb persistence.


Vyšlo v časopise: Reengineering Redox Sensitive GFP to Measure Mycothiol Redox Potential of during Infection. PLoS Pathog 10(1): e32767. doi:10.1371/journal.ppat.1003902
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1003902

Souhrn

Mycobacterium tuberculosis (Mtb) survives under oxidatively hostile environments encountered inside host phagocytes. To protect itself from oxidative stress, Mtb produces millimolar concentrations of mycothiol (MSH), which functions as a major cytoplasmic redox buffer. Here, we introduce a novel system for real-time imaging of mycothiol redox potential (EMSH) within Mtb cells during infection. We demonstrate that coupling of Mtb MSH-dependent oxidoreductase (mycoredoxin-1; Mrx1) to redox-sensitive GFP (roGFP2; Mrx1-roGFP2) allowed measurement of dynamic changes in intramycobacterial EMSH with unprecedented sensitivity and specificity. Using Mrx1-roGFP2, we report the first quantitative measurements of EMSH in diverse mycobacterial species, genetic mutants, and drug-resistant patient isolates. These cellular studies reveal, for the first time, that the environment inside macrophages and sub-vacuolar compartments induces heterogeneity in EMSH of the Mtb population. Further application of this new biosensor demonstrates that treatment of Mtb infected macrophage with anti-tuberculosis (TB) drugs induces oxidative shift in EMSH, suggesting that the intramacrophage milieu and antibiotics cooperatively disrupt the MSH homeostasis to exert efficient Mtb killing. Lastly, we analyze the membrane integrity of Mtb cells with varied EMSH during infection and show that subpopulation with higher EMSH are susceptible to clinically relevant antibiotics, whereas lower EMSH promotes antibiotic tolerance. Together, these data suggest the importance of MSH redox signaling in modulating mycobacterial survival following treatment with anti-TB drugs. We anticipate that Mrx1-roGFP2 will be a major contributor to our understanding of redox biology of Mtb and will lead to novel strategies to target redox metabolism for controlling Mtb persistence.


Zdroje

1. DyeC, WilliamsBG (2010) The population dynamics and control of tuberculosis. Science 328: 856–861.

2. EhrtS, SchnappingerD (2009) Mycobacterial survival strategies in the phagosome: defence against host stresses. Cell Microbiol 11: 1170–1178.

3. MacMickingJD, NorthRJ, LaCourseR, MudgettJS, ShahSK, et al. (1997) Identification of nitric oxide synthase as a protective locus against tuberculosis. Proc Natl Acad Sci U S A 94: 5243–5248.

4. CooperAM, SegalBH, FrankAA, HollandSM, OrmeIM (2000) Transient loss of resistance to pulmonary tuberculosis in p47(phox−/−) mice. Infect Immun 68: 1231–1234.

5. LeePP, ChanKW, JiangL, ChenT, LiC, et al. (2008) Susceptibility to mycobacterial infections in children with X-linked chronic granulomatous disease: a review of 17 patients living in a region endemic for tuberculosis. Pediatr Infect Dis J 27: 224–230.

6. YangCT, CambierCJ, DavisJM, HallCJ, CrosierPS, et al. (2012) Neutrophils exert protection in the early tuberculous granuloma by oxidative killing of mycobacteria phagocytosed from infected macrophages. Cell Host Microbe 12: 301–312.

7. den HengstCD, ButtnerMJ (2008) Redox control in actinobacteria. Biochim Biophys Acta 1780: 1201–1216.

8. BrugarolasP, MovahedzadehF, WangY, ZhangN, BartekIL, et al. (2012) The oxidation-sensing regulator (MosR) is a new redox-dependent transcription factor in Mycobacterium tuberculosis. J Biol Chem 287: 37703–37712.

9. SinghA, GuidryL, NarasimhuluKV, MaiD, TrombleyJ, et al. (2007) Mycobacterium tuberculosis WhiB3 responds to O2 and nitric oxide via its [4Fe-4S] cluster and is essential for nutrient starvation survival. Proc Natl Acad Sci U S A 104: 11562–11567.

10. SinghA, CrossmanDK, MaiD, GuidryL, VoskuilMI, et al. (2009) Mycobacterium tuberculosis WhiB3 maintains redox homeostasis by regulating virulence lipid anabolism to modulate macrophage response. PLoS Pathog 5: e1000545.

11. ChawlaM, ParikhP, SaxenaA, MunshiM, MehtaM, et al. (2012) Mycobacterium tuberculosis WhiB4 regulates oxidative stress response to modulate survival and dissemination in vivo. Mol Microbiol 85: 1148–1165.

12. SongT, DoveSL, LeeKH, HussonRN (2003) RshA, an anti-sigma factor that regulates the activity of the mycobacterial stress response sigma factor SigH. Mol Microbiol 50: 949–959.

13. FlynnJL, ChanJ (2001) Tuberculosis: latency and reactivation. Infect Immun 69: 4195–4201.

14. KumarA, FarhanaA, GuidryL, SainiV, HondalusM, et al. (2011) Redox homeostasis in mycobacteria: the key to tuberculosis control? Expert Rev Mol Med 13: e39.

15. RotaC, ChignellCF, MasonRP (1999) Evidence for free radical formation during the oxidation of 2′-7′-dichlorofluorescin to the fluorescent dye 2′-7′-dichlorofluorescein by horseradish peroxidase: possible implications for oxidative stress measurements. Free Radic Biol Med 27: 873–881.

16. TarpeyMM, WinkDA, GrishamMB (2004) Methods for detection of reactive metabolites of oxygen and nitrogen: in vitro and in vivo considerations. Am J Physiol Regul Integr Comp Physiol 286: R431–444.

17. MeyerAJ, DickTP (2010) Fluorescent protein-based redox probes. Antioxid Redox Signal 13: 621–650.

18. NewtonGL, RawatM, La ClairJJ, JothivasanVK, BudiartoT, et al. (2009) Bacillithiol is an antioxidant thiol produced in Bacilli. Nat Chem Biol 5: 625–627.

19. JothivasanVK, HamiltonCJ (2008) Mycothiol: synthesis, biosynthesis and biological functions of the major low molecular weight thiol in actinomycetes. Nat Prod Rep 25: 1091–1117.

20. SareenD, NewtonGL, FaheyRC, BuchmeierNA (2003) Mycothiol is essential for growth of Mycobacterium tuberculosis Erdman. J Bacteriol 185: 6736–6740.

21. BuchmeierNA, NewtonGL, KoledinT, FaheyRC (2003) Association of mycothiol with protection of Mycobacterium tuberculosis from toxic oxidants and antibiotics. Mol Microbiol 47: 1723–1732.

22. HansonGT, AggelerR, OglesbeeD, CannonM, CapaldiRA, et al. (2004) Investigating mitochondrial redox potential with redox-sensitive green fluorescent protein indicators. J Biol Chem 279: 13044–13053.

23. MeyerAJ, BrachT, MartyL, KreyeS, RouhierN, et al. (2007) Redox-sensitive GFP in Arabidopsis thaliana is a quantitative biosensor for the redox potential of the cellular glutathione redox buffer. Plant J 52: 973–986.

24. GutscherM, PauleauAL, MartyL, BrachT, WabnitzGH, et al. (2008) Real-time imaging of the intracellular glutathione redox potential. Nat Methods 5: 553–559.

25. Van LaerK, ButsL, FoloppeN, VertommenD, Van BelleK, et al. (2012) Mycoredoxin-1 is one of the missing links in the oxidative stress defence mechanism of Mycobacteria. Mol Microbiol 86: 787–804.

26. RawatM, NewtonGL, KoM, MartinezGJ, FaheyRC, et al. (2002) Mycothiol-deficient Mycobacterium smegmatis mutants are hypersensitive to alkylating agents, free radicals, and antibiotics. Antimicrob Agents Chemother 46: 3348–3355.

27. PatelMP, BlanchardJS (1999) Expression, purification, and characterization of Mycobacterium tuberculosis mycothione reductase. Biochemistry 38: 11827–11833.

28. Gutierrez-LugoMT, BakerH, ShiloachJ, BoshoffH, BewleyCA (2009) Dequalinium, a new inhibitor of Mycobacterium tuberculosis mycothiol ligase identified by high-throughput screening. J Biomol Screen 14: 643–652.

29. ArnerES, NakamuraH, SasadaT, YodoiJ, HolmgrenA, et al. (2001) Analysis of the inhibition of mammalian thioredoxin, thioredoxin reductase, and glutaredoxin by cis-diamminedichloroplatinum (II) and its major metabolite, the glutathione-platinum complex. Free Radic Biol Med 31: 1170–1178.

30. HaramakiN, HanD, HandelmanGJ, TritschlerHJ, PackerL (1997) Cytosolic and mitochondrial systems for NADH- and NADPH-dependent reduction of alpha-lipoic acid. Free Radic Biol Med 22: 535–542.

31. RawatM, JohnsonC, CadizV, Av-GayY (2007) Comparative analysis of mutants in the mycothiol biosynthesis pathway in Mycobacterium smegmatis. Biochem Biophys Res Commun 363: 71–76.

32. FernandesND, WuQL, KongD, PuyangX, GargS, et al. (1999) A mycobacterial extracytoplasmic sigma factor involved in survival following heat shock and oxidative stress. J Bacteriol 181: 4266–4274.

33. RamanS, SongT, PuyangX, BardarovS, JacobsWRJr, et al. (2001) The alternative sigma factor SigH regulates major components of oxidative and heat stress responses in Mycobacterium tuberculosis. J Bacteriol 183: 6119–6125.

34. KaushalD, SchroederBG, TyagiS, YoshimatsuT, ScottC, et al. (2002) Reduced immunopathology and mortality despite tissue persistence in a Mycobacterium tuberculosis mutant lacking alternative sigma factor, SigH. Proc Natl Acad Sci U S A 99: 8330–8335.

35. NewtonGL, TaP, FaheyRC (2005) A mycothiol synthase mutant of Mycobacterium smegmatis produces novel thiols and has an altered thiol redox status. J Bacteriol 187: 7309–7316.

36. TaP, BuchmeierN, NewtonGL, RawatM, FaheyRC (2011) Organic hydroperoxide resistance protein and ergothioneine compensate for loss of mycothiol in Mycobacterium smegmatis mutants. J Bacteriol 193: 1981–1990.

37. HolsclawCM, MuseWB3rd, CarrollKS, LearyJA (2011) Mass Spectrometric Analysis of Mycothiol levels in Wild-Type and Mycothiol Disulfide Reductase Mutant Mycobacterium smegmatis. Int J Mass Spectrom 305: 151–156.

38. AlbrechtSC, BarataAG, GrosshansJ, TelemanAA, DickTP (2011) In vivo mapping of hydrogen peroxide and oxidized glutathione reveals chemical and regional specificity of redox homeostasis. Cell Metab 14: 819–829.

39. UngKS, Av-GayY (2006) Mycothiol-dependent mycobacterial response to oxidative stress. FEBS Lett 580: 2712–2716.

40. ChanJ, XingY, MagliozzoRS, BloomBR (1992) Killing of virulent Mycobacterium tuberculosis by reactive nitrogen intermediates produced by activated murine macrophages. J Exp Med 175: 1111–1122.

41. ChanJ, TanakaK, CarrollD, FlynnJ, BloomBR (1995) Effects of nitric oxide synthase inhibitors on murine infection with Mycobacterium tuberculosis. Infect Immun 63: 736–740.

42. RohdeKH, VeigaDF, CaldwellS, BalazsiG, RussellDG (2012) Linking the transcriptional profiles and the physiological states of Mycobacterium tuberculosis during an extended intracellular infection. PLoS Pathog 8: e1002769.

43. LiuYB, LongMX, YinYJ, SiMR, ZhangL, et al. (2013) Physiological roles of mycothiol in detoxification and tolerance to multiple poisonous chemicals in Corynebacterium glutamicum. Arch Microbiol 195: 419–29.

44. RawatM, UppalM, NewtonG, SteffekM, FaheyRC, et al. (2004) Targeted mutagenesis of the Mycobacterium smegmatis mca gene, encoding a mycothiol-dependent detoxification protein. J Bacteriol 186: 6050–6058.

45. XuX, VilchezeC, Av-GayY, Gomez-VelascoA, JacobsWRJr (2011) Precise null deletion mutations of the mycothiol synthesis genes reveal their role in isoniazid and ethionamide resistance in Mycobacterium smegmatis. Antimicrob Agents Chemother 55: 3133–3139.

46. BrossierF, VezirisN, Truffot-PernotC, JarlierV, SougakoffW (2011) Molecular investigation of resistance to the antituberculous drug ethionamide in multidrug-resistant clinical isolates of Mycobacterium tuberculosis. Antimicrob Agents Chemother 55: 355–360.

47. YanoT, Kassovska-BratinovaS, TehJS, WinklerJ, SullivanK, et al. (2011) Reduction of clofazimine by mycobacterial type 2 NADH:quinone oxidoreductase: a pathway for the generation of bactericidal levels of reactive oxygen species. J Biol Chem 286: 10276–10287.

48. ConnollyLE, EdelsteinPH, RamakrishnanL (2007) Why is long-term therapy required to cure tuberculosis? PLoS Med 4: e120.

49. AdamsKN, TakakiK, ConnollyLE, WiedenhoftH, WingleeK, et al. (2011) Drug tolerance in replicating mycobacteria mediated by a macrophage-induced efflux mechanism. Cell 145: 39–53.

50. HewittCJ, Nebe-Von-CaronG (2001) An industrial application of multiparameter flow cytometry: assessment of cell physiological state and its application to the study of microbial fermentations. Cytometry 44: 179–187.

51. GutscherM, SobottaMC, WabnitzGH, BallikayaS, MeyerAJ, et al. (2009) Proximity-based protein thiol oxidation by H2O2-scavenging peroxidases. J Biol Chem 284: 31532–31540.

52. WelinA, RaffetsederJ, EklundD, StendahlO, LermM (2011) Importance of phagosomal functionality for growth restriction of Mycobacterium tuberculosis in primary human macrophages. J Innate Immun 3: 508–518.

53. ClemensDL, HorwitzMA (1996) The Mycobacterium tuberculosis phagosome interacts with early endosomes and is accessible to exogenously administered transferrin. J Exp Med 184: 1349–1355.

54. RussellDG, MwandumbaHC, RhoadesEE (2002) Mycobacterium and the coat of many lipids. J Cell Biol 158: 421–426.

55. HomolkaS, NiemannS, RussellDG, RohdeKH (2010) Functional genetic diversity among Mycobacterium tuberculosis complex clinical isolates: delineation of conserved core and lineage-specific transcriptomes during intracellular survival. PLoS Pathog 6: e1000988.

56. KooMS, SubbianS, KaplanG (2012) Strain specific transcriptional response in Mycobacterium tuberculosis infected macrophages. Cell Commun Signal 10: 2.

57. MancaC, PaulS, BarryCE3rd, FreedmanVH, KaplanG (1999) Mycobacterium tuberculosis catalase and peroxidase activities and resistance to oxidative killing in human monocytes in vitro. Infect Immun 67: 74–79.

58. IdhJ, MekonnenM, AbateE, WedajoW, WerngrenJ, et al. (2012) Resistance to first-line anti-TB drugs is associated with reduced nitric oxide susceptibility in Mycobacterium tuberculosis. PLoS One 7: e39891.

59. KohanskiMA, DwyerDJ, HayeteB, LawrenceCA, CollinsJJ (2007) A common mechanism of cellular death induced by bactericidal antibiotics. Cell 130: 797–810.

60. DwyerDJ, KohanskiMA, CollinsJJ (2009) Role of reactive oxygen species in antibiotic action and resistance. Curr Opin Microbiol 12: 482–489.

61. BrynildsenMP, WinklerJA, SpinaCS, MacdonaldIC, CollinsJJ (2013) Potentiating antibacterial activity by predictably enhancing endogenous microbial ROS production. Nat Biotechnol 31: 160–165.

62. KerenI, WuY, InocencioJ, MulcahyLR, LewisK (2013) Killing by bactericidal antibiotics does not depend on reactive oxygen species. Science 339: 1213–1216.

63. LiuY, ImlayJA (2013) Cell death from antibiotics without the involvement of reactive oxygen species. Science 339: 1210–1213.

64. Sao EmaniC, WilliamsMJ, WiidIJ, HitenNF, ViljoenAJ, et al. (2013) Ergothioneine is a secreted antioxidant in Mycobacterium smegmatis. Antimicrob Agents Chemother 57L: 3202–7.

65. KimJJ, LeeHM, ShinDM, KimW, YukJM, et al. (2012) Host cell autophagy activated by antibiotics is required for their effective antimycobacterial drug action. Cell Host Microbe 11: 457–468.

66. VilchezeC, Av-GayY, AttarianR, LiuZ, HazbonMH, et al. (2008) Mycothiol biosynthesis is essential for ethionamide susceptibility in Mycobacterium tuberculosis. Mol Microbiol 69: 1316–1329.

67. RussellDG, VanderVenBC, LeeW, AbramovitchRB, KimMJ, et al. (2010) Mycobacterium tuberculosis wears what it eats. Cell Host Microbe 8: 68–76.

68. ViaLE, LinPL, RaySM, CarrilloJ, AllenSS, et al. (2008) Tuberculous granulomas are hypoxic in guinea pigs, rabbits, and nonhuman primates. Infect Immun 76: 2333–2340.

69. BoshoffHI, XuX, TahlanK, DowdCS, PetheK, et al. (2008) Biosynthesis and recycling of nicotinamide cofactors in Mycobacterium tuberculosis. An essential role for NAD in nonreplicating bacilli. J Biol Chem 283: 19329–19341.

70. LinPL, DartoisV, JohnstonPJ, JanssenC, ViaL, et al. (2012) Metronidazole prevents reactivation of latent Mycobacterium tuberculosis infection in macaques. Proc Natl Acad Sci U S A 109: 14188–14193.

71. GrantSS, KaufmannBB, ChandNS, HaseleyN, HungDT (2012) Eradication of bacterial persisters with antibiotic-generated hydroxyl radicals. Proc Natl Acad Sci U S A 109: 12147–12152.

72. FortuneSM (2012) The Surprising Diversity of Mycobacterium tuberculosis: Change You Can Believe In. J Infect Dis 206: 1642–1644.

73. RankinS, LiZ, IsbergRR (2002) Macrophage-induced genes of Legionella pneumophila: protection from reactive intermediates and solute imbalance during intracellular growth. Infect Immun 70: 3637–3648.

74. SinghA, MaiD, KumarA, SteynAJ (2006) Dissecting virulence pathways of Mycobacterium tuberculosis through protein-protein association. Proc Natl Acad Sci U S A 103: 11346–11351.

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2014 Číslo 1
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#